Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Transl Med ; 17(1): 219, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31288845

RESUMEN

BACKGROUND: The pleiotropic cytokine, transforming growth factor (TGF)-ß, and CD4+CD25+Foxp3+ regulatory T cells (Tregs) play a critical role in actively suppressing antitumor immune responses. Evidence shows that TGF-ß produced by tumor cells promotes tolerance via expansion of Tregs. Our group previously demonstrated that blockade of TGF-ß signaling with a small molecule TGF-ß receptor I antagonist (SM16) inhibited primary and metastatic tumor growth in a T cell dependent fashion. In the current study, we evaluated the effect of SM16 on Treg generation and function. METHODS: Using BALB/c, FoxP3eGFP and Rag-/- mice, we performed FACS analysis to determine if SM16 blocked de novo TGF-ß-induced Treg generation in vitro and in vivo. CD4+ T cells from lymph node and spleen were isolated from control mice or mice maintained on SM16 diet, and flow cytometry analysis was used to detect the frequency of CD4+CD25-FoxP3+ and CD4+CD25+FoxP3+ T cells. In vitro suppression assays were used to determine the ability to suppress naive T cell proliferation in vitro of both CD4+CD25+FoxP3+ and CD4+CD25-FoxP3+ T cell sub-populations. We then examined whether SM16 diet exerted an inhibitory effect on primary tumor growth and correlated with changes in FoxP3+expression. ELISA analysis was used to measure IFN-γ levels after 72 h co-culture of CD4+CD25+ T cells from tumor-bearing mice on control or SM16 diet with CD4+CD25- T cells from naive donors. RESULTS: SM16 abrogates TGF-ß-induced Treg generation in vitro but does not prevent global homeostatic expansion of CD4+ T cell sub-populations in vivo. Instead, SM16 treatment causes expansion of a population of CD4+CD25-Foxp3+ Treg-like cells without significantly altering the overall frequency of Treg in lymphoreplete naive and tumor-bearing mice. Importantly, both the CD4+CD25-Foxp3+ T cells and the CD4+CD25+Foxp3+ Tregs in mice receiving SM16 diet exhibited diminished ability to suppress naive T cell proliferation in vitro compared to Treg from mice on control diet. CONCLUSIONS: These findings suggest that blockade of TGF-ß signaling is a potentially useful strategy for blunting Treg function to enhance the anti-tumor response. Our data further suggest that the overall dampening of Treg function may involve the expansion of a quiescent Treg precursor population, which is CD4+CD25-Foxp3+.


Asunto(s)
Antineoplásicos/metabolismo , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Recuento de Células , Línea Celular Tumoral , Proliferación Celular , Femenino , Homeostasis , Interferón gamma/metabolismo , Ganglios Linfáticos/citología , Ratones Noqueados , Neoplasias/patología , Bazo/citología , Linfocitos T Reguladores/inmunología , Carga Tumoral
2.
Int J Cancer ; 143(12): 3201-3208, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30248181

RESUMEN

Like in many tumor types, immunotherapy is currently under investigation to assess its potential efficacy in glioblastoma patients. Trials are under way to assess the efficacy of new immune checkpoint inhibitors including anti-PD-1 or CTLA4. We here investigate the expression and efficacy of a novel immune-checkpoint inhibitor, called LAG-3. We show that LAG-3 is expressed in human glioblastoma samples and in a mouse glioblastoma model we show that knock out or LAG-3 inhibition with a blocking antibody is efficacious against glioblastoma and can be used in combination with other immune checkpoint inhibitors toward complete eradication of the model glioblastoma tumors. From a mechanistic standpoint we show that LAG-3 expression is an early marker of T cell exhaustion and therefore early treatment with LAG-3 blocking antibody is more efficacious than later treatment. These data provide insight and support the design of trials that incorporate LAG-3 in the treatment of glioblastoma.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Receptor de Muerte Celular Programada 1/inmunología , Anciano , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos CD/genética , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Femenino , Citometría de Flujo , Glioblastoma/inmunología , Humanos , Inmunohistoquímica , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína del Gen 3 de Activación de Linfocitos
3.
Ann N Y Acad Sci ; 1089: 343-72, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17261780

RESUMEN

The extensive literature and the work from our laboratory illustrate the large number of complex processes affected by estrogen that might contribute to the striking ability of 17-beta estradiol (E2) and its derivatives to inhibit clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) in mice. These effects require sustained exposure to relatively low doses of exogenous hormone and offer better protection when initiated prior to induction of EAE. The E2 mediates inhibition of encephalitogenic T cells, inhibition of cell migration into central nervous system tissue, and neuroprotective effects that promote axon and myelin survival. E2 effects on EAE are mediated through Esr-1 (alpha receptor for E2) but not Esr-2 (beta receptor for E2), as are its anti-inflammatory and neuroprotective effects. A novel finding is that E2 upregulated the expression of FoxP3 that contributes to the activity of CD4 + CD25 + T regulatory cells (Treg). The protective effects of E2 in EAE suggest its use as a therapy for multiple sclerosis (MS). Possible risks may be minimized by using sub-pregnancy levels of exogenous E2 that produced synergistic effects when used in combination with another immunoregulatory therapy. Alternatively, one might envision using E2 derivatives alone or in combination therapies in both male and female MS patients.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Estradiol/farmacología , Estrógenos/fisiología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Estradiol/uso terapéutico , Humanos , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Linfocitos T Reguladores/inmunología
4.
Sci Transl Med ; 8(370): 370ra180, 2016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-28003545

RESUMEN

The immunosuppressive effects of chemotherapy present a challenge for designing effective cancer immunotherapy strategies. We hypothesized that although systemic chemotherapy (SC) exhibits negative immunologic effects, local chemotherapy (LC) can potentiate an antitumor immune response. We show that LC combined with anti-programmed cell death protein 1 (PD-1) facilitates an antitumor immune response and improves survival (P < 0.001) in glioblastoma. LC-treated mice had increased infiltration of tumor-associated dendritic cells and clonal expansion of antigen-specific T effector cells. In comparison, SC resulted in systemic and intratumoral lymphodepletion, with decreased immune memory in long-term survivors. Furthermore, adoptive transfer of CD8+ cells from LC-treated mice partially rescued SC-treated mice after tumor rechallenge. Last, the timing of chemo- and immunotherapy had differential effects on anti-PD-1 efficacy. This study suggests that both mode of delivery and timing have distinct effects on the efficacy of anti-PD-1. The results of this work could help guide the selection and scheduling of combination treatment for patients with glioblastoma and other tumor types.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/inmunología , Animales , Carmustina/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Glioma/tratamiento farmacológico , Humanos , Inmunosupresores/farmacología , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Resultado del Tratamiento
5.
J Neuroimmunol ; 170(1-2): 85-92, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16253347

RESUMEN

Estrogen (E2) upregulates the FoxP3 gene that marks regulatory CD4+CD25+ T cells (Treg cells). However, E2 also inhibits the ability of antigen presenting cells (APC) to activate T cells. It is possible that these opposing functions might affect the degree of overt suppression during pregnancy and autoimmunity. To evaluate E2 effects on Treg cell function, we quantified FoxP3 levels and Treg suppression in CD4+CD25+ T cells from pregnant and E2-treated mice, and overt Treg suppression in E2- vs. placebo-pretreated mice with autoimmune encephalomyelitis. The data clearly demonstrate that enhanced expression of FoxP3, which occurs in pregnant mice and in mice treated exogenously with E2 pellets, results in a concomitant increase in functional suppression within the CD4+CD25(bright) Treg fraction of splenocytes. The similarities in FoxP3 expression and Treg cell function in E2-treated and pregnant mice implicate E2 as a major contributor for increasing Treg function during pregnancy. Surprisingly, suppression was not enhanced when Treg cells from E2-treated mice were activated with APC and CD4+CD25- responder T cells from the same E2-treated mice, a result consistent with impaired APC activation of Treg cells. In contrast, Treg suppression was strikingly enhanced in combined cell cultures from E2-pretreated mice that were protected from EAE induced with neuroantigen in complete Freund's adjuvant. These results suggest that E2 treatment may have opposing effects on Treg cells vs. APC that both contribute to overt suppression, but such effects are overcome and focused towards enhanced suppression in inflammatory environments produced during pregnancy and EAE.


Asunto(s)
Estradiol/farmacología , Factores de Transcripción Forkhead/metabolismo , Preñez/fisiología , Linfocitos T Reguladores/fisiología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Embarazo , Preñez/metabolismo , Receptores de Interleucina-2/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Células Th2/metabolismo
6.
Int Immunol ; 19(3): 337-43, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17267414

RESUMEN

Estrogen [17-beta-estradiol (E2)] is a potent driver of the FoxP3+ regulatory T cell (Treg) compartment. Recently, Tregs were further characterized by intracellular expression of the negative co-stimulatory molecule, programmed death-1 (PD-1). To clarify the role of PD-1 versus FoxP3 in E2-enhanced Treg suppression, we evaluated both markers and functional suppression in wild-type, estrogen receptor knockout (ERKO) mice and PD-1 KO mice. We demonstrate that intracellular PD-1 expression is also E2 sensitive, since E2 treatment increased intracellular PD-1 levels in CD4+FoxP3+ cells, and PD-1 expression and Treg suppression were reduced in ERKO mice. Surprisingly, PD-1 KO mice retained normal levels of FoxP3 expression, but Tregs from these mice lacked functional suppression. However, E2 pre-treatment of PD-1 KO mice partially restored functional Treg suppression without enhancing FoxP3 expression. Thus, functional Treg suppression in immunized mice without E2 pre-treatment was more closely linked to PD-1 expression than to FoxP3 expression. However, although enhanced PD-1 expression was E2 dependent, functional suppression was still enhanced by E2 pre-treatment in the absence of PD-1. These data clearly demonstrate that E2 can affect multiple regulatory elements that influence Treg suppression, including both PD-1-dependent and PD-1-independent pathways.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Estradiol/metabolismo , Factores de Transcripción Forkhead/metabolismo , Receptores de Estrógenos/deficiencia , Linfocitos T Reguladores/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Antígenos/inmunología , Antígenos de Superficie/genética , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Estradiol/farmacología , Receptor alfa de Estrógeno/deficiencia , Receptor beta de Estrógeno/deficiencia , Femenino , Glicoproteínas/inmunología , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/inmunología , Receptor de Muerte Celular Programada 1 , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
7.
J Neurosci Res ; 84(2): 370-8, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16676326

RESUMEN

Estrogen (E2)-induced immunomodulation involves dual effects on antigen-presenting cells (APC) and CD4(+)CD25(+) regulatory T cells (Treg) but not a direct effect on effector T cells. In this report, we further investigated the effects of E2 on APC and Treg function. We found that E2 treatment in vivo strongly reduced recovery of APC from the peritoneal cavity and inhibited induction of the inflammatory cytokines interleukin (IL)-12 and interferon-gamma but enhanced secretion of IL-10. Moreover, E2-conditioned bone marrow-derived dendritic cells (BM-DC) could both enhance Treg activity and directly inhibit responder T cells in the absence of Treg cells. We examined whether this E2-induced inhibitory activity of BM-DC might involve costimulation through the recently described PD-1 pathway. Both E2 and pregnancy markedly enhanced PD-1 expression in several types of APC, including macrophages, B cells, and especially dendritic cells (DC). Similarly to E2-induced enhancement of FoxP3 expression and experimental autoimmune encephalomyelitis protection, E2-induced enhancement of PD-1(+) cells was also mediated through estrogen receptor alpha (Esr1) in DC and macrophages but not in B cells. Based on antibody inhibition studies, PD-1 interaction with its ligands, PDL-1 and especially PDL-2, could mediate either positive or negative regulatory signaling in both mature and immature E2-conditioned DC, depending, respectively, on a relatively high (10:1) or low (1:1) ratio of T cells:BM-DC. These novel findings indicate that E2-induced immunomodulation is mediated in part through potentiation in BM-DC of the PD-1 costimulatory pathway.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Estrógenos/metabolismo , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Antígenos de Diferenciación/efectos de los fármacos , Medios de Cultivo Condicionados , Femenino , Factores Inmunológicos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Ratones , Receptor de Muerte Celular Programada 1 , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
8.
J Gen Virol ; 86(Pt 7): 2007-2018, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15958680

RESUMEN

BALB/c mice infected with the Moscow strain of Ectromelia virus (ECTV-MOS) show a large number of apoptotic cells, and an influx of lymphoid cells in the epithelium and substantia propria of conjunctivae, respectively. The presence of ECTV-MOS antigens in the epithelium of conjunctivae significantly upregulates Fas in the epithelial layer and FasL in the suprabasal layer of conjunctiva. Inhibition of FasL with blocking antibodies in cultures of conjunctival cells isolated from ECTV-MOS-infected BALB/c mice showed that the Fas/FasL pathway is important in apoptosis of ECTV-MOS-infected cells. The results also showed that the presence of cytokines, in particular interferon (IFN)-gamma, upregulated expression of Fas. Interleukin (IL) 2, 4, 10 and IFN-gamma were produced at the peak of conjunctivitis (at day 15 of infection) with a predominance of IFN-gamma and a small, but significant, production of IL4 and IL10 compared with non-infected animals. These results suggest that not only is Fas/FasL expression in conjunctiva involved in elimination of migrating Fas+ cells but also plays an important role in the turnover of conjunctival epithelium and thus may be crucial for ECTV spreading to the surrounding environment.


Asunto(s)
Apoptosis , Conjuntivitis Viral/fisiopatología , Virus de la Ectromelia/patogenicidad , Células Epiteliales/fisiología , Glicoproteínas de Membrana/metabolismo , Receptor fas/metabolismo , Animales , Conjuntiva/citología , Conjuntiva/patología , Conjuntiva/virología , Conjuntivitis Viral/inmunología , Conjuntivitis Viral/virología , Citocinas/metabolismo , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/fisiopatología , Ectromelia Infecciosa/virología , Células Epiteliales/virología , Proteína Ligando Fas , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Regulación hacia Arriba
9.
J Immunol ; 173(4): 2227-30, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15294932

RESUMEN

CD4(+)CD25(+) regulatory T cells are crucial to the maintenance of tolerance in normal individuals. However, the factors regulating this cell population and its function are largely unknown. Estrogen has been shown to protect against the development of autoimmune disease, yet the mechanism is not known. We demonstrate that estrogen (17-beta-estradiol, E2) is capable of augmenting FoxP3 expression in vitro and in vivo. Treatment of naive mice with E2 increased both CD25(+) cell number and FoxP3 expression level. Further, the ability of E2 to protect against autoimmune disease (experimental autoimmune encephalomyelitis) correlated with its ability to up-regulate FoxP3, as both were reduced in estrogen receptor alpha-deficient animals. Finally, E2 treatment and pregnancy induced FoxP3 protein expression to a similar degree, suggesting that high estrogen levels during pregnancy may help to maintain fetal tolerance. In summary, our data suggest E2 promotes tolerance by expanding the regulatory T cell compartment.


Asunto(s)
Antígenos CD4/inmunología , Estrógenos/inmunología , Tolerancia Inmunológica , Receptores de Interleucina-2/inmunología , Linfocitos T/inmunología , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Receptor alfa de Estrógeno , Estrógenos/farmacología , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead , Activación de Linfocitos/inmunología , Ratones , Embarazo , Receptores de Estrógenos/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/efectos de los fármacos
10.
Am J Pathol ; 165(6): 2069-77, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15579449

RESUMEN

Gender influences mediated by 17 beta-estradiol (E2) have been associated with susceptibility to and severity of autoimmune diseases such as diabetes, arthritis, and multiple sclerosis. In this regard, we have shown that estrogen receptor-alpha (Esr1) is crucial for the protective effect of 17 beta-estradiol (E2) in murine experimental autoimmune encephalitis (EAE), an animal model of multiple sclerosis. The expression of estrogen receptors among various immune cells (eg, T and B lymphocytes, antigen-presenting cells) suggests that the therapeutic effect of E2 is likely mediated directly through specific receptor binding. However, the target immune cell populations responsive to E2 treatment have not been identified. In the current study, we induced EAE in T-cell-deficient, severe combined immunodeficient mice or in immunocompetent mice with encephalitogenic T cells from wild-type Esr1+/+ or Esr1 knockout (Esr1-/-) donors and compared the protective E2 responses. The results showed that E2-responsive, Esr1+/+ disease-inducing encephalitogenic T cells were neither necessary nor sufficient for E2-mediated protection from EAE. Instead, the therapeutic response appeared to be mediated through direct effects on nonlymphocytic, E2-responsive cells and down-regulation of the inflammatory response in the central nervous system. These results provide the first demonstration that the protective effect of E2 on EAE is not mediated directly through E2-responsive T cells and raise the alternative possibility that nonlymphocytic cells such as macrophages, dendritic cells, or other nonlymphocytic cells are primarily responsive to E2 treatment in EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/fisiología , Linfocitos T , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/inmunología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/inmunología , Femenino , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
11.
Clin Immunol ; 110(2): 181-90, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15003815

RESUMEN

Dehydroepiandrosterone (DHEA), a precursor of immune-regulating hormones (IRH) including the androstenes, has attracted much interest over the last several decades because of its many antiaging, metabolic, and immune modulating effects. 5-Androstene-16alpha fluoro-17-one (fluasterone, also known as HE2500) is a synthetic androstene analogue that retains anti-inflammatory, antiproliferative, and immune-regulating activities of the parent molecule, but is nontoxic and practically devoid of androgenic or estrogenic side effects. In the present studies, we tested the ability of fluasterone to limit disease in the DBA mouse model of collagen-induced arthritis (CIA). We found that mice receiving injections of fluasterone displayed significant delay in onset, decrease in CIA peak score, and significant decrease of the daily mean clinical score. Benefit was associated with significant decreases in (1). bovine type II collagen (bCII)-specific IgG(1) and IgG(2a) antibody levels in serum; (2). production of TNF-alpha, IL-6, IFN-gamma, but not IL-10; (3). lymphocyte proliferative response to bCII protein; and (4). joint inflammation, erosion, and synovial proliferation as judged by histological analysis. This is the first study to report that an IRH can ameliorate ongoing disease in a CIA mouse model with relevance to RA and to correlate that finding with decreases in pro-inflammatory cytokines.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Deshidroepiandrosterona/análogos & derivados , Deshidroepiandrosterona/farmacología , Animales , Artritis Experimental/patología , División Celular/efectos de los fármacos , División Celular/inmunología , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Histocitoquímica , Inmunoglobulina G/sangre , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
12.
Am J Pathol ; 163(4): 1599-605, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14507666

RESUMEN

Low-dose estrogen (E2) treatment significantly inhibits the clinical signs and histopathological lesions of experimental autoimmune encephalomyelitis (EAE), and is being used in clinical trials to treat multiple sclerosis. To assess the role of intracytoplasmic estrogen receptors in mediating suppression of EAE, we studied mice with disrupted estrogen receptor-alpha (Esr1) and -beta (Esr2) genes. We demonstrate that the protective effect of E2 is abrogated in B6.129-Esr1(tm1Unc) mice (Esr1-/-) but not in B6.129-Esr2(tm1Unc) mice (Esr2-/-). The loss of E2-mediated protection from EAE in Esr1-/- mice immunized with the encephalitogenic MOG-35-55 peptide was manifested phenotypically by the development of severe acute clinical signs and histopathological lesions even in the presence of moderately high serum E2 levels. This is in contrast to C57BL/6 wild-type (WT) mice and Esr2-/- mice in which E2 treatment resulted in comparable serum levels and markedly suppressed clinical signs of EAE and abolished inflammatory lesions in the CNS. This pattern showing a lack of E2-dependent inhibition of EAE in Esr1-/- mice was mirrored by an enhanced rather than a reduced secretion of TNF-alpha, IFN-gamma, and interleukin (IL)-6 in MOG-specific splenocytes and a lack of inhibition of message for inflammatory cytokines, chemokines and chemokine receptors in CNS tissue. These results indicate that the immunomodulatory effects of E2 in EAE are dependent on Esr1 and not Esr2 signaling.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Estradiol/farmacología , Receptores de Estrógenos/metabolismo , Animales , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Quimiocina/biosíntesis
13.
Am J Pathol ; 164(6): 1915-24, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15161628

RESUMEN

Estrogens and estrogen-receptor signaling function in establishing and regulating the female immune system and it is becoming increasingly evident that they may play a similar role in males. We report that B10.PL/SnJ male mice with a disrupted estrogen receptor-1 (alpha) gene (Esr1(-/-)) develop less severe clinical experimental allergic encephalomyelitis (EAE) compared to either Esr1(+/-) or wild-type (Esr1(+/+)) controls when immunized with myelin basic protein peptide Ac1-11 (MBP(Ac1-11)). In contrast, the disease course in B10.PL/SnJ male mice with a disrupted estrogen receptor-2 (beta) gene (Esr2(-/-)) does not differ from that of wild-type (Esr2(+/+)) mice. However, Esr2(+/-) mice do develop more severe clinical disease with an earlier onset indicating that heterosis at Esr2 plays a significant role in regulating EAE in males. No significant differences in central nervous system histopathology or MBP(Ac1-11)-specific T-cell responses as assessed by proliferation and interleukin-2 production were observed as a function of either Esr1 or Esr2 genotype. An analysis of cytokine/chemokine secretion by MBP(Ac1-11)-specific T cells revealed unique Esr1 and Esr2 genotype-dependent regulation. Interferon-gamma secretion was found to be negatively regulated by Esr1 whereas interleukin-6 and tumor necrosis factor-alpha secretion exhibited classical Esr2 gene dose responses. Interestingly, MCP-1 displayed distinctively unique patterns of genotype-dependent regulation by Esr1 and Esr2. The contribution of the hematopoietic and nonhematopoietic cellular compartments associated with the heterotic effect at Esr2 in regulating the severity of clinical EAE was identified using reciprocal hematopoietic radiation bone marrow chimeras generated between male wild-type and Esr2(+/-) mice. Wild-type --> Esr2(+/-) mice exhibited EAE equivalent in severity to that seen in Esr2(+/-) --> Esr2(+/-) control constructs; both of which were more severe than the clinical signs observed in Esr2(+/-) --> wild-type and wild-type --> wild-type mice. These results indicate that the heterotic effect at Esr2 is a function of the nonhematopoietic compartment.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Receptores de Estrógenos/fisiología , Animales , Médula Ósea/patología , Encéfalo/patología , División Celular , Quimera , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA